Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Rev Panam Salud Publica ; 48: e71, 2024.
Article in Spanish | MEDLINE | ID: mdl-39286658

ABSTRACT

The Region of the Americas has historically experienced social inequalities rooted in colonialism, which are reflected and reproduced in the area of health. The COVID-19 pandemic affected the entire Region, but the most socially disadvantaged groups were hit hardest, intensifying health inequities. Under the premise that pandemics are not socially neutral phenomena, this special report analyzes the unequal impacts of the pandemic from different perspectives: historical, epidemiological, political, social, economic, environmental, and population-related. Critical reflections are offered here on the negative impacts of inequalities on well-being, not only in the most affected populations, but across society as a whole. Strategic recommendations are made for progress toward health equity in the post-pandemic context. This report highlights the importance of advancing toward mature information systems to monitor health equity, developing more resilient health systems, and implementing explicit policies and practices aimed at eliminating health inequities. All of this should pave the way for prosperity and sustainable development in the Region.


Historicamente, a Região das Américas vivencia desigualdades sociais enraizadas no colonialismo, que estão refletidas e se reproduzem no campo da saúde. A pandemia de COVID-19 afetou toda a Região, mas atingiu com mais força os grupos mais desfavorecidos do ponto de vista social, agravando as iniquidades em saúde. Sob a premissa de que as pandemias não são fenômenos neutros em termos sociais, este relatório especial analisa os impactos desiguais da pandemia a partir de diferentes perspectivas: histórica, epidemiológica, política, social, econômica, ambiental e populacional. São apresentadas reflexões críticas sobre as implicações negativas das desigualdades para o bem-estar, não apenas das populações mais afetadas, mas da sociedade como um todo. Conclui-se com recomendações estratégicas para avançar em direção à equidade em saúde no cenário pós-pandemia. Destaca-se a importância de avançar na maturidade dos sistemas de informação para monitorar a equidade em saúde, a resiliência dos sistemas de saúde e a implementação de políticas e práticas explícitas voltadas para a eliminação das iniquidades em saúde. Espera-se que os pontos mencionados abram caminho para a prosperidade e o desenvolvimento sustentável na Região.

2.
Genes Dev ; 30(12): 1440-53, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27340176

ABSTRACT

Superenhancers (SEs) are large genomic regions with a high density of enhancer marks. In cancer, SEs are found near oncogenes and dictate cancer gene expression. However, how oncogenic SEs are regulated remains poorly understood. Here, we show that INO80, a chromatin remodeling complex, is required for SE-mediated oncogenic transcription and tumor growth in melanoma. The expression of Ino80, the SWI/SNF ATPase, is elevated in melanoma cells and patient melanomas compared with normal melanocytes and benign nevi. Furthermore, Ino80 silencing selectively inhibits melanoma cell proliferation, anchorage-independent growth, tumorigenesis, and tumor maintenance in mouse xenografts. Mechanistically, Ino80 occupies >90% of SEs, and its occupancy is dependent on transcription factors such as MITF and Sox9. Ino80 binding reduces nucleosome occupancy and facilitates Mediator recruitment, thus promoting oncogenic transcription. Consistently, genes co-occupied by Ino80 and Med1 are selectively expressed in melanomas compared with melanocytes. Together, our results reveal an essential role of INO80-dependent chromatin remodeling in SE function and suggest a novel strategy for disrupting SEs in cancer treatment.


Subject(s)
Carcinogenesis/genetics , Cell Cycle Proteins/metabolism , Enhancer Elements, Genetic/physiology , Gene Expression Regulation, Neoplastic/genetics , Melanoma/genetics , Melanoma/physiopathology , Nuclear Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Proliferation/genetics , Chromatin Assembly and Disassembly/genetics , Gene Silencing , Heterografts , Humans , Mediator Complex Subunit 1/genetics , Melanocytes/metabolism , Melanoma/enzymology , Mice , Nuclear Proteins/genetics , Protein Binding , Transcription Factors/metabolism
3.
Int J Equity Health ; 22(1): 121, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37381010

ABSTRACT

BACKGROUND: The enduring threat of maternal mortality to health worldwide and in the Americas has been recognized in the global and regional agendas and their targets to 2030. To inform the direction and amount of effort needed to meet those targets, a set of equity-sensitive regional scenarios of maternal mortality ratio (MMR) reduction based on its tempo or speed of change from baseline year 2015 was developed. METHODS: Regional scenarios by 2030 were defined according to: i) the MMR average annual rate of reduction (AARR) needed to meet the global (70 per 100,000) or regional (30 per 100,000) targets and, ii) the horizontal (proportional) or vertical (progressive) equity criterion applied to the cross-country AARR distribution (i.e., same speed to all countries or faster for those with higher baseline MMR). MMR average and inequality gaps -absolute (AIG), and relative (RIG)- were scenario outcomes. RESULTS: At baseline, MMR was 59.2 per 100,000; AIG was 313.4 per 100,000 and RIG was 19.0 between countries with baseline MMR over twice the global target and those below the regional target. The AARR needed to meet the global and regional targets were -7.60% and -4.54%, respectively; baseline AARR was -1.55%. In the regional MMR target attainment scenario, applying horizontal equity would decrease AIG to 158.7 per 100,000 and RIG will remain invariant; applying vertical equity would decrease AIG to 130.9 per 100,000 and RIG would decrease to 13.5 by 2030. CONCLUSION: The dual challenge of reducing maternal mortality and abating its inequalities will demand hefty efforts from countries of the Americas. This remains true to their collective 2030 MMR target while leaving no one behind. These efforts should be mainly directed towards significantly speeding up the tempo of the MMR reduction and applying sensible progressivity, targeting on groups and territories with higher MMR and greater social vulnerabilities, especially in a post-pandemic regional context.


Subject(s)
Maternal Mortality , Humans , Americas/epidemiology , Female
4.
J Biol Chem ; 297(5): 101335, 2021 11.
Article in English | MEDLINE | ID: mdl-34688654

ABSTRACT

Oncogenic KRAS drives cancer growth by activating diverse signaling networks, not all of which have been fully delineated. We set out to establish a system-wide profile of the KRAS-regulated kinase signaling network (kinome) in KRAS-mutant pancreatic ductal adenocarcinoma (PDAC). We knocked down KRAS expression in a panel of six cell lines and then applied multiplexed inhibitor bead/MS to monitor changes in kinase activity and/or expression. We hypothesized that depletion of KRAS would result in downregulation of kinases required for KRAS-mediated transformation and in upregulation of other kinases that could potentially compensate for the deleterious consequences of the loss of KRAS. We identified 15 upregulated and 13 downregulated kinases in common across the panel of cell lines. In agreement with our hypothesis, all 15 of the upregulated kinases have established roles as cancer drivers (e.g., SRC, TGF-ß1, ILK), and pharmacological inhibition of one of these upregulated kinases, DDR1, suppressed PDAC growth. Interestingly, 11 of the 13 downregulated kinases have established driver roles in cell cycle progression, particularly in mitosis (e.g., WEE1, Aurora A, PLK1). Consistent with a crucial role for the downregulated kinases in promoting KRAS-driven proliferation, we found that pharmacological inhibition of WEE1 also suppressed PDAC growth. The unexpected paradoxical activation of ERK upon WEE1 inhibition led us to inhibit both WEE1 and ERK concurrently, which caused further potent growth suppression and enhanced apoptotic death compared with WEE1 inhibition alone. We conclude that system-wide delineation of the KRAS-regulated kinome can identify potential therapeutic targets for KRAS-mutant pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal , Cell Cycle Proteins/metabolism , MAP Kinase Signaling System/drug effects , Mutation , Pancreatic Neoplasms , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins p21(ras) , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
5.
Bull World Health Organ ; 100(1): 40-49, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-35017756

ABSTRACT

OBJECTIVE: To assess the availability and gaps in data for measuring progress towards health-related sustainable development goals and other targets in selected low- and middle-income countries. METHODS: We used 14 international population surveys to evaluate the health data systems in the 47 least developed countries over the years 2015-2020. We reviewed the survey instruments to determine whether they contained tools that could be used to measure 46 health-related indicators defined by the World Health Organization. We recorded the number of countries with data available on the indicators from these surveys. FINDINGS: Twenty-seven indicators were measurable by the surveys we identified. The two health emergency indicators were not measurable by current surveys. The percentage of countries that used surveys to collect data over 2015-2020 were lowest for tuberculosis (2/47; 4.3%), hepatitis B (3/47; 6.4%), human immunodeficiency virus (11/47; 23.4%), child development status and child abuse (both 13/47; 27.7%), compared with safe drinking water (37/47; 78.7%) and births attended by skilled health personnel (36/47; 76.6%). Nineteen countries collected data on 21 or more indicators over 2015-2020 while nine collected data on no indicators; over 2018-2020 these numbers reduced to six and 20, respectively. CONCLUSION: Examining selected international surveys provided a quick summary of health data available in the 47 least developed countries. We found major gaps in health data due to long survey cycles and lack of appropriate survey instruments. Novel indicators and survey instruments would be needed to track the fast-changing situation of health emergencies.


Subject(s)
Developing Countries , Goals , Child , Humans , Income , Sustainable Development , World Health Organization
6.
Semin Cell Dev Biol ; 58: 60-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27422332

ABSTRACT

Mutationally activated RAS proteins are critical oncogenic drivers in nearly 30% of all human cancers. As with mutant RAS, the role of wild type RAS proteins in oncogenesis, tumour maintenance and metastasis is context-dependent. Complexity is introduced by the existence of multiple RAS genes (HRAS, KRAS, NRAS) and protein "isoforms" (KRAS4A, KRAS4B), by the ever more complicated network of RAS signaling, and by the increasing identification of numerous genetic aberrations in cancers that do and do not harbour mutant RAS. Numerous mouse model carcinogenesis studies and examination of patient tumours reveal that, in RAS-mutant cancers, wild type RAS proteins are likely to serve as tumour suppressors when the mutant RAS is of the same isoform. This evidence is particularly robust in KRAS mutant cancers, which often display suppression or loss of wild type KRAS, but is not as strong for NRAS. In contrast, although not yet fully elucidated, the preponderance of evidence indicates that wild type RAS proteins play a tumour promoting role when the mutant RAS is of a different isoform. In non-RAS mutant cancers, wild type RAS is recognized as a mediator of oncogenic signaling due to chronic activation of upstream receptor tyrosine kinases that feed through RAS. Additionally, in the absence of mutant RAS, activation of wild type RAS may drive cancer upon the loss of negative RAS regulators such as NF1 GAP or SPRY proteins. Here we explore the current state of knowledge with respect to the roles of wild type RAS proteins in human cancers.


Subject(s)
Neoplasms/metabolism , ras Proteins/metabolism , Amino Acid Sequence , Animals , Humans , Models, Biological , Mutation/genetics , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction , ras Proteins/chemistry , ras Proteins/genetics
7.
Proc Natl Acad Sci U S A ; 112(3): 779-84, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25561545

ABSTRACT

The two products of the KRAS locus, K-Ras4A and K-Ras4B, are encoded by alternative fourth exons and therefore, possess distinct membrane-targeting sequences. The common activating mutations occur in exons 1 or 2 and therefore, render both splice variants oncogenic. K-Ras4A has been understudied, because it has been considered a minor splice variant. By priming off of the splice junction, we developed a quantitative RT-PCR assay for K-Ras4A and K-Ras4B message capable of measuring absolute amounts of the two transcripts. We found that K-Ras4A was widely expressed in 30 of 30 human cancer cell lines and amounts equal to K-Ras4B in 17 human colorectal tumors. Using splice variant-specific antibodies, we detected K-Ras4A protein in several tumor cell lines at a level equal to or greater than that of K-Ras4B. In addition to the CAAX motif, the C terminus of K-Ras4A contains a site of palmitoylation as well as a bipartite polybasic region. Although both were required for maximal efficiency, each of these could independently deliver K-Ras4A to the plasma membrane. Thus, among four Ras proteins, K-Ras4A is unique in possessing a dual membrane-targeting motif. We also found that, unlike K-Ras4B, K-Ras4A does not bind to the cytosolic chaperone δ-subunit of cGMP phosphodiesterase type 6 (PDE6δ). We conclude that efforts to develop anti-K-Ras drugs that interfere with membrane trafficking will have to take into account the distinct modes of targeting of the two K-Ras splice variants.


Subject(s)
Genes, ras , Neoplasms/genetics , RNA Splicing , Amino Acid Sequence , Cell Line, Tumor , Humans , Molecular Sequence Data , Polymerase Chain Reaction
8.
J Biol Chem ; 291(34): 17804-15, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27226552

ABSTRACT

The protein kinase casein kinase 2 (CK2) is a pleiotropic and constitutively active kinase that plays crucial roles in cellular proliferation and survival. Overexpression of CK2, particularly the α catalytic subunit (CK2α, CSNK2A1), has been implicated in a wide variety of cancers and is associated with poorer survival and resistance to both conventional and targeted anticancer therapies. Here, we found that CK2α protein is elevated in melanoma cell lines compared with normal human melanocytes. We then tested the involvement of CK2α in drug resistance to Food and Drug Administration-approved single agent targeted therapies for melanoma. In BRAF mutant melanoma cells, ectopic CK2α decreased sensitivity to vemurafenib (BRAF inhibitor), dabrafenib (BRAF inhibitor), and trametinib (MEK inhibitor) by a mechanism distinct from that of mutant NRAS. Conversely, knockdown of CK2α sensitized cells to inhibitor treatment. CK2α-mediated RAF-MEK kinase inhibitor resistance was tightly linked to its maintenance of ERK phosphorylation. We found that CK2α post-translationally regulates the ERK-specific phosphatase dual specificity phosphatase 6 (DUSP6) in a kinase dependent-manner, decreasing its abundance. However, we unexpectedly showed, by using a kinase-inactive mutant of CK2α, that RAF-MEK inhibitor resistance did not rely on CK2α kinase catalytic function, and both wild-type and kinase-inactive CK2α maintained ERK phosphorylation upon inhibition of BRAF or MEK. That both wild-type and kinase-inactive CK2α bound equally well to the RAF-MEK-ERK scaffold kinase suppressor of Ras 1 (KSR1) suggested that CK2α increases KSR facilitation of ERK phosphorylation. Accordingly, CK2α did not cause resistance to direct inhibition of ERK by the ERK1/2-selective inhibitor SCH772984. Our findings support a kinase-independent scaffolding function of CK2α that promotes resistance to RAF- and MEK-targeted therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Extracellular Signal-Regulated MAP Kinases , MAP Kinase Signaling System , Melanoma , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Dual Specificity Phosphatase 6/genetics , Dual Specificity Phosphatase 6/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Melanoma/drug therapy , Melanoma/enzymology , Melanoma/genetics , Melanoma/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism
9.
J Biol Chem ; 290(37): 22851-61, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26216878

ABSTRACT

The Ras-like small GTPases RalA and RalB are well validated effectors of RAS oncogene-driven human cancer growth, and pharmacologic inhibitors of Ral function may provide an effective anti-Ras therapeutic strategy. Intriguingly, although RalA and RalB share strong overall amino acid sequence identity, exhibit essentially identical structural and biochemical properties, and can utilize the same downstream effectors, they also exhibit divergent and sometimes opposing roles in the tumorigenic and metastatic growth of different cancer types. These distinct biological functions have been attributed largely to sequence divergence in their carboxyl-terminal hypervariable regions. However, the role of posttranslational modifications signaled by the hypervariable region carboxyl-terminal tetrapeptide CAAX motif (C = cysteine, A = aliphatic amino acid, X = terminal residue) in Ral isoform-selective functions has not been addressed. We determined that these modifications have distinct roles and consequences. Both RalA and RalB require Ras converting CAAX endopeptidase 1 (RCE1) for association with the plasma membrane, albeit not with endomembranes, and loss of RCE1 caused mislocalization as well as sustained activation of both RalA and RalB. In contrast, isoprenylcysteine carboxylmethyltransferase (ICMT) deficiency disrupted plasma membrane localization only of RalB, whereas RalA depended on ICMT for efficient endosomal localization. Furthermore, the absence of ICMT increased stability of RalB but not RalA protein. Finally, palmitoylation was critical for subcellular localization of RalB but not RalA. In summary, we have identified striking isoform-specific consequences of distinct CAAX-signaled posttranslational modifications that contribute to the divergent subcellular localization and activity of RalA and RalB.


Subject(s)
Protein Processing, Post-Translational/physiology , ral GTP-Binding Proteins/metabolism , Amino Acid Motifs , Animals , Cell Membrane/genetics , Cell Membrane/metabolism , Endosomes/genetics , Endosomes/metabolism , Humans , Mice , Mice, Knockout , Protein Transport/physiology , ral GTP-Binding Proteins/genetics
10.
Cancer Cell ; 12(5): 467-78, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17996650

ABSTRACT

Chronic myelogenous leukemia (CML) is a clonal myeloproliferative disease (MPD) initiated by expression of the p210-BCR-ABL fusion protein. We demonstrate in a murine model of p210-BCR-ABL-induced MPD that gene targeting of Rac1 and Rac2 significantly delays or abrogates disease development. Attenuation of the disease phenotype is associated with severely diminished p210-BCR-ABL-induced downstream signaling in primary hematopoietic cells. We utilize NSC23766, a small molecule antagonist of Rac activation, to validate biochemically and functionally Rac as a molecular target in both a relevant animal model and in primary human CML cells in vitro and in a xenograft model in vivo, including in Imatinib-resistant p210-BCR-ABL disease. These data demonstrate that Rac is an additional therapeutic target in p210-BCR-ABL-mediated MPD.


Subject(s)
Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , rac GTP-Binding Proteins/physiology , Aminoquinolines/pharmacology , Animals , Antigens, CD34/biosynthesis , Cell Line, Tumor , Dose-Response Relationship, Drug , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Mice , Myeloproliferative Disorders/therapy , Neoplasm Transplantation , Phenotype , Pyrimidines/pharmacology , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , RAC2 GTP-Binding Protein
11.
J Biol Chem ; 288(46): 33156-70, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24089523

ABSTRACT

Histone deacetylase 6 (HDAC6) is well known for its ability to promote cell migration through deacetylation of its cytoplasmic substrates such as α-tubulin. However, how HDAC6 itself is regulated to control cell motility remains elusive. Previous studies have shown that one third of extracellular signal-regulated kinase (ERK) is associated with the microtubule cytoskeleton in cells. Yet, no connection between HDAC6 and ERK has been discovered. Here, for the first time, we reveal that ERK binds to and phosphorylates HDAC6 to promote cell migration via deacetylation of α-tubulin. We have identified two novel ERK-mediated phosphorylation sites: threonine 1031 and serine 1035 in HDAC6. Both sites were phosphorylated by ERK1 in vitro, whereas Ser-1035 was phosphorylated in response to the activation of EGFR-Ras-Raf-MEK-ERK signaling pathway in vivo. HDAC6-null mouse embryonic fibroblasts rescued by the nonphosphorylation mimicking mutant displayed significantly reduced cell migration compared with those rescued by the wild type. Consistently, the nonphosphorylation mimicking mutant exerted lower tubulin deacetylase activity in vivo compared with the wild type. These data indicate that ERK/HDAC6-mediated cell motility is through deacetylation of α-tubulin. Overall, our results suggest that HDAC6-mediated cell migration could be governed by EGFR-Ras-Raf-MEK-ERK signaling.


Subject(s)
Cell Movement/physiology , Histone Deacetylases/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Tubulin/metabolism , Acetylation , Animals , CHO Cells , Cricetinae , Cricetulus , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Histone Deacetylase 6 , Histone Deacetylases/genetics , Humans , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinase 3/genetics , Tubulin/genetics
12.
Science ; 385(6715): 1274-1275, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39298611

ABSTRACT

A single small molecule degrades numerous KRAS variants involved in cancer.

13.
Science ; 384(6700): eadk0850, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843329

ABSTRACT

To delineate the mechanisms by which the ERK1 and ERK2 mitogen-activated protein kinases support mutant KRAS-driven cancer growth, we determined the ERK-dependent phosphoproteome in KRAS-mutant pancreatic cancer. We determined that ERK1 and ERK2 share near-identical signaling and transforming outputs and that the KRAS-regulated phosphoproteome is driven nearly completely by ERK. We identified 4666 ERK-dependent phosphosites on 2123 proteins, of which 79 and 66%, respectively, were not previously associated with ERK, substantially expanding the depth and breadth of ERK-dependent phosphorylation events and revealing a considerably more complex function for ERK in cancer. We established that ERK controls a highly dynamic and complex phosphoproteome that converges on cyclin-dependent kinase regulation and RAS homolog guanosine triphosphatase function (RHO GTPase). Our findings establish the most comprehensive molecular portrait and mechanisms by which ERK drives KRAS-dependent pancreatic cancer growth.


Subject(s)
Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Pancreatic Neoplasms , Phosphoproteins , Proteome , Proto-Oncogene Proteins p21(ras) , Animals , Humans , Mice , Cell Line, Tumor , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/genetics , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Phosphoproteins/metabolism , Phosphoproteins/genetics , Phosphorylation , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , HEK293 Cells
14.
Science ; 384(6700): eadk0775, 2024 06 07.
Article in English | MEDLINE | ID: mdl-38843331

ABSTRACT

How the KRAS oncogene drives cancer growth remains poorly understood. Therefore, we established a systemwide portrait of KRAS- and extracellular signal-regulated kinase (ERK)-dependent gene transcription in KRAS-mutant cancer to delineate the molecular mechanisms of growth and of inhibitor resistance. Unexpectedly, our KRAS-dependent gene signature diverges substantially from the frequently cited Hallmark KRAS signaling gene signature, is driven predominantly through the ERK mitogen-activated protein kinase (MAPK) cascade, and accurately reflects KRAS- and ERK-regulated gene transcription in KRAS-mutant cancer patients. Integration with our ERK-regulated phospho- and total proteome highlights ERK deregulation of the anaphase promoting complex/cyclosome (APC/C) and other components of the cell cycle machinery as key processes that drive pancreatic ductal adenocarcinoma (PDAC) growth. Our findings elucidate mechanistically the critical role of ERK in driving KRAS-mutant tumor growth and in resistance to KRAS-ERK MAPK targeted therapies.


Subject(s)
Carcinoma, Pancreatic Ductal , Extracellular Signal-Regulated MAP Kinases , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Mutation , Pancreatic Neoplasms , Proto-Oncogene Proteins p21(ras) , Transcriptome , Animals , Humans , Mice , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , HEK293 Cells
15.
J Biol Chem ; 287(18): 14827-36, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22393054

ABSTRACT

Ras-like (Ral) small GTPases are regulated downstream of Ras and the noncanonical Ral guanine nucleotide exchange factor (RalGEF) effector pathway. Despite RalA and RalB sharing 82% sequence identity and utilization of shared effector proteins, their roles in normal and neoplastic cell growth have been shown to be highly distinct. Here, we determined that RalB function is regulated by protein kinase Cα (PKCα) phosphorylation. We found that RalB phosphorylation on Ser-198 in the C-terminal membrane targeting sequence resulted in enhanced RalB endomembrane accumulation and decreased RalB association with its effector, the exocyst component Sec5. Additionally, RalB phosphorylation regulated vesicular trafficking and membrane fusion by regulating v- and t-SNARE interactions. RalB phosphorylation regulated vesicular traffic of α5-integrin to the cell surface and cell attachment to fibronectin. In summary, our data suggest that phosphorylation by PKCα is critical for RalB-mediated vesicle trafficking and exocytosis.


Subject(s)
Exocytosis/physiology , Protein Kinase C-alpha/metabolism , ral GTP-Binding Proteins/metabolism , Animals , Cell Line , Enzyme Activation/physiology , Humans , Phosphorylation/physiology , Protein Kinase C-alpha/genetics , Protein Transport/physiology , Rats , SNARE Proteins/genetics , SNARE Proteins/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , ral GTP-Binding Proteins/genetics
16.
Cancer Cell ; 7(6): 533-45, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15950903

ABSTRACT

RalGEFs were recently shown to be critical for Ras-mediated transformed and tumorigenic growth of human cells. We now show that the oncogenic activity of these proteins is propagated by activation of one RalGEF substrate, RalA, but blunted by another closely related substrate, RalB, and that the oncogenic signaling requires binding of the RalBP1 and exocyst subunit effector proteins. Knockdown of RalA expression impeded, if not abolished, the ability of human cancer cells to form tumors. RalA was also commonly activated in a panel of cell lines from pancreatic cancers, a disease characterized by activation of Ras. Activation of RalA signaling thus appears to be a critical step in Ras-induced transformation and tumorigenesis of human cells.


Subject(s)
Cell Transformation, Neoplastic/pathology , Proto-Oncogene Proteins p21(ras)/physiology , ral GTP-Binding Proteins/physiology , ATP-Binding Cassette Transporters/metabolism , Animals , Carrier Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , GTPase-Activating Proteins/metabolism , Gene Expression/genetics , Guanosine Triphosphate/metabolism , Humans , Mice , Mice, SCID , Neoplasm Transplantation/pathology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Binding/physiology , Protein Transport/physiology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Small Interfering/genetics , Transfection , Vesicular Transport Proteins , ral GTP-Binding Proteins/genetics , ral GTP-Binding Proteins/metabolism , ral Guanine Nucleotide Exchange Factor/genetics , ral Guanine Nucleotide Exchange Factor/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
17.
Cancer Discov ; 13(1): 19-22, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36620884

ABSTRACT

SUMMARY: In this issue, Hattori and colleagues capitalized on targeted small-molecule covalent inhibitors of one KRAS mutant with a G12C substitution and of other oncoproteins to create drug-peptide conjugates that serve as cancer neoantigens that prompt an immune response to oncogene-mutant cancer cells. This immunotherapy strategy can serve as an effective approach to overcome the treatment-induced resistance that limits the effectiveness of essentially all small molecule-based targeted anticancer drugs. See related article by Hattori et al., p. 132 (9).


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Oncogenes , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry
18.
Genes Cancer ; 14: 30-49, 2023.
Article in English | MEDLINE | ID: mdl-36923647

ABSTRACT

We and others have recently shown that proteins involved in the DNA damage response (DDR) are critical for KRAS-mutant pancreatic ductal adenocarcinoma (PDAC) cell growth in vitro. However, the CRISPR-Cas9 library that enabled us to identify these key proteins had limited representation of DDR-related genes. To further investigate the DDR in this context, we performed a comprehensive, DDR-focused CRISPR-Cas9 loss-of-function screen. This screen identified valosin-containing protein (VCP) as an essential gene in KRAS-mutant PDAC cell lines. We observed that genetic and pharmacologic inhibition of VCP limited cell growth and induced apoptotic death. Addressing the basis for VCP-dependent growth, we first evaluated the contribution of VCP to the DDR and found that loss of VCP resulted in accumulation of DNA double-strand breaks. We next addressed its role in proteostasis and found that loss of VCP caused accumulation of polyubiquitinated proteins. We also found that loss of VCP increased autophagy. Therefore, we reasoned that inhibiting both VCP and autophagy could be an effective combination. Accordingly, we found that VCP inhibition synergized with the autophagy inhibitor chloroquine. We conclude that concurrent targeting of autophagy can enhance the efficacy of VCP inhibitors in KRAS-mutant PDAC.

19.
Cancer Res ; 83(24): 4112-4129, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37934103

ABSTRACT

Primary/intrinsic and treatment-induced acquired resistance limit the initial response rate to and long-term efficacy of direct inhibitors of the KRASG12C mutant in cancer. To identify potential mechanisms of resistance, we applied a CRISPR/Cas9 loss-of-function screen and observed loss of multiple components of the Hippo tumor suppressor pathway, which acts to suppress YAP1/TAZ-regulated gene transcription. YAP1/TAZ activation impaired the antiproliferative and proapoptotic effects of KRASG12C inhibitor (G12Ci) treatment in KRASG12C-mutant cancer cell lines. Conversely, genetic suppression of YAP1/WWTR1 (TAZ) enhanced G12Ci sensitivity. YAP1/TAZ activity overcame KRAS dependency through two distinct TEAD transcription factor-dependent mechanisms, which phenocopy KRAS effector signaling. First, TEAD stimulated ERK-independent transcription of genes normally regulated by ERK (BIRC5, CDC20, ECT2, FOSL1, and MYC) to promote progression through the cell cycle. Second, TEAD caused activation of PI3K-AKT-mTOR signaling to overcome apoptosis. G12Ci treatment-induced acquired resistance was also caused by YAP1/TAZ-TEAD activation. Accordingly, concurrent treatment with pharmacologic inhibitors of TEAD synergistically enhanced KRASG12C inhibitor antitumor activity in vitro and prolonged tumor suppression in vivo. In summary, these observations reveal YAP1/TAZ-TEAD signaling as a crucial driver of primary and acquired resistance to KRAS inhibition and support the use of TEAD inhibitors to enhance the antitumor efficacy of KRAS-targeted therapies. SIGNIFICANCE: YAP1/TAZ-TEAD activation compensates for loss of KRAS effector signaling, establishing a mechanistic basis for concurrent inhibition of TEAD to enhance the efficacy of KRASG12C-selective inhibitor treatment of KRASG12C-mutant cancers. See related commentary by Johnson and Haigis, p. 4005.


Subject(s)
Drug Resistance, Neoplasm , Neoplasms , TEA Domain Transcription Factors , Humans , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Trans-Activators/metabolism , YAP-Signaling Proteins , TEA Domain Transcription Factors/antagonists & inhibitors
20.
Cancer Res ; 83(1): 141-157, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36346366

ABSTRACT

Mutational loss of CDKN2A (encoding p16INK4A) tumor-suppressor function is a key genetic step that complements activation of KRAS in promoting the development and malignant growth of pancreatic ductal adenocarcinoma (PDAC). However, pharmacologic restoration of p16INK4A function with inhibitors of CDK4 and CDK6 (CDK4/6) has shown limited clinical efficacy in PDAC. Here, we found that concurrent treatment with both a CDK4/6 inhibitor (CDK4/6i) and an ERK-MAPK inhibitor (ERKi) synergistically suppresses the growth of PDAC cell lines and organoids by cooperatively blocking CDK4/6i-induced compensatory upregulation of ERK, PI3K, antiapoptotic signaling, and MYC expression. On the basis of these findings, a Phase I clinical trial was initiated to evaluate the ERKi ulixertinib in combination with the CDK4/6i palbociclib in patients with advanced PDAC (NCT03454035). As inhibition of other proteins might also counter CDK4/6i-mediated signaling changes to increase cellular CDK4/6i sensitivity, a CRISPR-Cas9 loss-of-function screen was conducted that revealed a spectrum of functionally diverse genes whose loss enhanced CDK4/6i growth inhibitory activity. These genes were enriched around diverse signaling nodes, including cell-cycle regulatory proteins centered on CDK2 activation, PI3K-AKT-mTOR signaling, SRC family kinases, HDAC proteins, autophagy-activating pathways, chromosome regulation and maintenance, and DNA damage and repair pathways. Novel therapeutic combinations were validated using siRNA and small-molecule inhibitor-based approaches. In addition, genes whose loss imparts a survival advantage were identified (e.g., RB1, PTEN, FBXW7), suggesting possible resistance mechanisms to CDK4/6 inhibition. In summary, this study has identified novel combinations with CDK4/6i that may have clinical benefit to patients with PDAC. SIGNIFICANCE: CRISPR-Cas9 screening and protein activity mapping reveal combinations that increase potency of CDK4/6 inhibitors and overcome drug-induced compensations in pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Pancreatic Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL